1
|
Wowui PI, Mprah R, Ndzie Noah ML, Adu-Amankwaah J, Kanoseh AWL, Tao L, Chulu D, Yalley SK, Shaheen S, Sun H. Estrogen via GPER downregulated HIF-1a and MIF expression, attenuated cardiac arrhythmias, and myocardial inflammation during hypobaric hypoxia. Mol Med 2025; 31:107. [PMID: 40108505 PMCID: PMC11924608 DOI: 10.1186/s10020-025-01144-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/27/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND The human body is highly dependent on adequate oxygenation of the cellular space for physiologic homeostasis mediation. The insufficient oxygenation of the cellular space leads to hypoxia. Hypobaric hypoxia (HH) is the reduction in oxygen partial pressure and atmospheric pressure during ascent to high altitudes. This state induces a maladaptive response. Women and how hormones like estrogen influence hypoxia have not been explored with most research being conducted on males. In this study, we investigated the effects of estrogen and GPER on HIF-1a and MIF expression, cardiac arrhythmias, and inflammation during hypobaric hypoxia. METHODS Ovariectomy and SHAM operations were done on FVB wild-type (WT) female mice. 2 weeks after the operation, the mice were treated with estrogen (40 mg/kg) as a therapeutic intervention and placed in a hypoxic chamber at an altitude of 6000 m for 7 days. Cardiac electrical activity was assessed using electrocardiography. Alterations in protein expression, inflammatory, and GPER pathways were investigated using western blotting, ELISA, and immunofluorescence. Histological assessment was performed using Masson's trichrome staining. Peritoneal macrophages were isolated for in vitro study. RESULTS Under hypobaric hypoxia (HH), the ovariectomized (OVX) group showed increased macrophage migration inhibitory factor (MIF) and hypoxia-inducible factor-1 alpha (HIF-1α) expression. In contrast, these factors were downregulated in the estrogen-treated and control groups. HH also caused cardiac inflammation and fibrosis, especially in the OVX + HH group, which had elevated proinflammatory cytokines (IL-1β, IL-6, TNF-α) and decreased anti-inflammatory cytokines (TGF-β, IL-10). Inhibition with G15 (a GPER antagonist) increased MIF and HIF-1α, whereas activation with G1 (a GPER agonist) decreased their expression, highlighting GPER's crucial role in regulating MIF during HH. CONCLUSION Estrogen regulates HIF-1α and MIF expression through the GPER during hypobaric hypoxia, suggesting a potential therapeutic pathway to mitigate maladaptive responses during high-altitude ascent.
Collapse
Affiliation(s)
- Prosperl Ivette Wowui
- Department of Physiology, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Richard Mprah
- Department of Physiology, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Marie Louise Ndzie Noah
- Department of Physiology, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Joseph Adu-Amankwaah
- Department of Physiology, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | | | - Li Tao
- Department of Physiology, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Diana Chulu
- Department of Physiology, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Simon Kumah Yalley
- Department of Physiology, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Saffia Shaheen
- Department of Physiology, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Hong Sun
- Department of Physiology, School of Basic Medical Sciences, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China.
- Xuzhou Key Laboratory of Physiological Function and Injury, Xuzhou Medical University, Xuzhou, China.
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
2
|
Bojko B, Claudia S, Vladimir V, Aleksandra K, Stefan S, Marko J, Ilija B, Maciej B. The Relationship Between Ambulatory Blood Pressure and Atherogenic Index with LV Geometry Pattern in Obese Children: Results from a Cross-Sectional Study in Serbia. Pediatr Cardiol 2024:10.1007/s00246-024-03749-6. [PMID: 39729282 DOI: 10.1007/s00246-024-03749-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024]
Abstract
Arterial hypertension and increased atherogenic index of plasma (AIP) are strong predictors of cardiovascular risk associated in individuals with obesity both in adults and children. Thus, we aimed to explore the relationship between AIP and systolic ambulatory blood pressure index (sABPI) with left ventricular geometry pattern in obese children. In this cross-sectional study, a total of 129 obese children (BMI greater or equal to the 95th percentile for age and sex) were examined. Fasting blood samples were taken to measure plasma glucose level, lipid profile, including total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), triglycerides (TG), serum uric acid (SUA), and insulin level. Two-dimensional (2-D) transthoracic echocardiography was performed to determine left ventricular geometry pattern. Ambulatory blood pressure recording was obtained for 24 h in all obese children. Linear regression analyses were conducted to explore relationships between AIP (calculated as logarithmic transformation of the ratio of TG to HDL-C) and sABPI with left ventricular myocardial mass index (LVMI) and relative wall thickness (RWT). We also used sex as a potential effect modifier, and calculated stratum-specific estimates of the effect. We demonstrated independent and positive association of age and AIP with RWT (Age: effect size = 0.83 (CI 0.22 - 1.45) p = 0.008; AI effect size 8.9 (CI 3.6 - 14.3); p = 0.01). In a subgroup analysis with sex as an effect modifier, independent positive association of borderline significance at p = 0.011 was found between AIP and RWT only in boys AIP: effect size 9.5 (CI 2.3 - 16.7) p = 0.01. LVMI was significantly associated with sex and BMI (sex: effect size = 6.8 CI (2.6 - 11) p = 0.002; BMI z score: effect size = 6.8 CI (3.2-10.4) p < 0.001). Independent positive association was also found between BMI and LVMI for girls, effect size = 11.9 (CI 4.1-19.8) (p = 0.005). AIP and age are independently associated with the presence of concentric left ventricular geometry with girls seeming more protected from atherogenic hypertrophic stimulus than boys. BMI and sex are independently associated with eccentric left ventricular remodeling, with BMI being a greater risk factor for girls.
Collapse
Affiliation(s)
- Bjelakovic Bojko
- Clinic of Pediatrics, Clinical Center, Nis Zorana Djindjica 48 Boulevard, 18000, Nis, Serbia.
- Medical Faculty, University of Nis, Nis, Serbia.
| | - Stefanutti Claudia
- Extracorporeal Therapeutic Techniques Unit, Regional Centre for Rare Disease Immunohematology and Transfusion Medicine, Department of Molecular Medicine, Lipid Clinic and Atherosclerosis Prevention Centre, ''Sapienza'' University of Rome, ''Umberto I'' Hospital, Rome, Italy
| | - Vukovic Vladimir
- Center for Disease Control and Prevention, Institute of Public Health of Vojvodina, Novi Sad, Serbia
| | - Klisic Aleksandra
- Primary Health Center, Podgorica, Montenegro
- Faculty of Medicine, University of Montenegro, Podgorica, Montenegro
| | - Stojkovic Stefan
- Clinic for Gastroenterohepatology, University Clinical Center of Serbia, Beograd, Serbia
- Cicarrone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jovic Marko
- Medical Faculty, University of Nis, Nis, Serbia
| | | | - Banach Maciej
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland
- Faculty of Medicine, the John Paul II Catholic University of Lublin, Lublin, Poland
- Cicarrone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
3
|
Gerges SH, El-Kadi AOS. Changes in cardiovascular arachidonic acid metabolism in experimental models of menopause and implications on postmenopausal cardiac hypertrophy. Prostaglandins Other Lipid Mediat 2024; 173:106851. [PMID: 38740361 DOI: 10.1016/j.prostaglandins.2024.106851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 04/17/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024]
Abstract
Menopause is a normal stage in the human female aging process characterized by the cessation of menstruation and the ovarian production of estrogen and progesterone hormones. Menopause is associated with an increased risk of several different diseases. Cardiovascular diseases are generally less common in females than in age-matched males. However, this female advantage is lost after menopause. Cardiac hypertrophy is a disease characterized by increased cardiac size that develops as a response to chronic overload or stress. Similar to other cardiovascular diseases, the risk of cardiac hypertrophy significantly increases after menopause. However, the exact underlying mechanisms are not yet fully elucidated. Several studies have shown that surgical or chemical induction of menopause in experimental animals is associated with cardiac hypertrophy, or aggravates cardiac hypertrophy induced by other stressors. Arachidonic acid (AA) released from the myocardial phospholipids is metabolized by cardiac cytochrome P450 (CYP), cyclooxygenase (COX), and lipoxygenase (LOX) enzymes to produce several eicosanoids. AA-metabolizing enzymes and their respective metabolites play an important role in the pathogenesis of cardiac hypertrophy. Menopause is associated with changes in the cardiovascular levels of CYP, COX, and LOX enzymes and the levels of their metabolites. It is possible that these changes might play a role in the increased risk of cardiac hypertrophy after menopause.
Collapse
Affiliation(s)
- Samar H Gerges
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
4
|
Vetter TA, Parthiban P, Stevens JA, Revelo XS, Kohr MJ, Townsend D. Reduced cardiac antioxidant defenses mediate increased susceptibility to workload-induced myocardial injury in males with genetic cardiomyopathy. J Mol Cell Cardiol 2024; 190:24-34. [PMID: 38527667 PMCID: PMC11060907 DOI: 10.1016/j.yjmcc.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 03/27/2024]
Abstract
Ongoing cardiomyocyte injury is a major mechanism in the progression of heart failure, particularly in dystrophic hearts. Due to the poor regenerative capacity of the adult heart, cardiomyocyte death results in the permanent loss of functional myocardium. Understanding the factors contributing to myocyte injury is essential for the development of effective heart failure therapies. As a model of persistent cardiac injury, we examined mice lacking β-sarcoglycan (β-SG), a key component of the dystrophin glycoprotein complex (DGC). The loss of the sarcoglycan complex markedly compromises sarcolemmal integrity in this β-SG-/- model. Our studies aim to characterize the mechanisms underlying dramatic sex differences in susceptibility to cardiac injury in β-SG-/- mice. Male β-SG-/- hearts display significantly greater myocardial injury and death following isoproterenol-induced cardiac stress than female β-SG-/- hearts. This protection of females was independent of ovarian hormones. Male β-SG-/- hearts displayed increased susceptibility to exogenous oxidative stress and were significantly protected by angiotensin II type 1 receptor (AT1R) antagonism. Increasing general antioxidative defenses or increasing the levels of S-nitrosylation both provided protection to the hearts of β-SG-/- male mice. Here we demonstrate that increased susceptibility to oxidative damage leads to an AT1R-mediated amplification of workload-induced myocardial injury in male β-SG-/- mice. Improving oxidative defenses, specifically by increasing S-nitrosylation, provided protection to the male β-SG-/- heart from workload-induced injury. These studies describe a unique susceptibility of the male heart to injury and may contribute to the sex differences in other forms of cardiac injury.
Collapse
Affiliation(s)
- Tatyana A Vetter
- Nationwide Children's Hospital, Columbus, OH, United States of America
| | - Preethy Parthiban
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Jackie A Stevens
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Xavier S Revelo
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Mark J Kohr
- Department of Environmental Health and Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - DeWayne Townsend
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States of America; Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota, Minneapolis, MN, United States of America; Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States of America.
| |
Collapse
|
5
|
Pawlicki P, Yurdakok-Dikmen B, Tworzydlo W, Kotula-Balak M. Toward understanding the role of the interstitial tissue architects: Possible functions of telocytes in the male gonad. Theriogenology 2024; 217:25-36. [PMID: 38241912 DOI: 10.1016/j.theriogenology.2024.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 01/21/2024]
Abstract
Telocytes represent a relatively recently discovered population of interstitial cells with a unique morphological structure that distinguishes them from other neighboring cells. Through their long protrusions extending from the cell body, telocytes create microenvironments via tissue compartmentalization and create homo- and hetero-cellular junctions. These establish a three-dimensional network enabling the maintenance of interstitial compartment homeostasis through regulation of extracellular matrix organization and activity, structural support, paracrine and juxtracrine communication, immunomodulation, immune surveillance, cell survival, and apoptosis. The presence of telocytes has also been confirmed in testicular interstitial tissue of many species of animals. The objective of this review is to summarize recent findings on telocytes in the male gonad, on which conclusions have been deduced that indicate the involvement of telocytes in maintaining the cytoarchitecture of the testicular interstitial tissue, in the processes of spermatogenesis and steroidogenesis, and photoperiod-mediated changes in the testes in seasonally reproductive animals.
Collapse
Affiliation(s)
- Piotr Pawlicki
- Center of Experimental and Innovative Medicine, University of Agriculture in Krakow, Redzina 1c, 30-248, Krakow, Poland.
| | - Begum Yurdakok-Dikmen
- Department of Pharmacology and Toxicology, Ankara University Faculty of Veterinary Medicine, Ankara, 06110, Dışkapı, Turkey.
| | - Waclaw Tworzydlo
- Department of Developmental Biology and Invertebrate Morphology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, Gronostajowa 9, 30-385, Krakow, Poland.
| | - Malgorzata Kotula-Balak
- Department of Animal Anatomy and Preclinical Sciences, University Centre of Veterinary Medicine JU-UA, University of Agriculture in Krakow, Mickiewicza 24/28, 30-059, Krakow, Poland.
| |
Collapse
|
6
|
Taube N, Kabir R, Ebenebe OV, Garbus H, Alam El Din SM, Illingworth E, Fitch M, Wang N, Kohr MJ. Prenatal arsenite exposure alters maternal cardiac remodeling during late pregnancy. Toxicol Appl Pharmacol 2024; 483:116833. [PMID: 38266874 PMCID: PMC10922692 DOI: 10.1016/j.taap.2024.116833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 01/08/2024] [Accepted: 01/21/2024] [Indexed: 01/26/2024]
Abstract
Exposure to inorganic arsenic through drinking water is widespread and has been linked to many chronic diseases, including cardiovascular disease. Arsenic exposure has been shown to alter hypertrophic signaling in the adult heart, as well as in utero offspring development. However, the effect of arsenic on maternal cardiac remodeling during pregnancy has not been studied. As such, there is a need to understand how environmental exposure contributes to adverse pregnancy-related cardiovascular events. This study seeks to understand the impact of trivalent inorganic arsenic exposure during gestation on maternal cardiac remodeling in late pregnancy, as well as offspring outcomes. C57BL/6 J mice were exposed to 0 (control), 100 or 1000 μg/L sodium arsenite (NaAsO2) beginning at embryonic day (E) 2.5 and continuing through E17.5. Maternal heart function and size were assessed via transthoracic echocardiography, gravimetric measurement, and histology. Transcript levels of hypertrophic markers were probed via qRT-PCR and confirmed by western blot. Offspring outcomes were assessed through echocardiography and gravimetric measurement. We found that maternal heart size was smaller and transcript levels of Esr1 (estrogen receptor alpha), Pgrmc1 (progesterone receptor membrane component 1) and Pgrmc2 (progesterone receptor membrane component 2) reduced during late pregnancy with exposure to 1000 μg/L iAs vs. non-exposed pregnant controls. Both 100 and 1000 μg/L iAs also reduced transcription of Nppa (atrial natriuretic peptide). Akt protein expression was also significantly reduced after 1000 μg/L iAs exposure in the maternal heart with no change in activating phosphorylation. This significant abrogation of maternal cardiac hypertrophy suggests that arsenic exposure during pregnancy can potentially contribute to cardiovascular disease. Taken together, our findings further underscore the importance of reducing arsenic exposure during pregnancy and indicate that more research is needed to assess the impact of arsenic and other environmental exposures on the maternal heart and adverse pregnancy events.
Collapse
Affiliation(s)
- Nicole Taube
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Raihan Kabir
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Obialunanma V Ebenebe
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Haley Garbus
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Sarah-Marie Alam El Din
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Emily Illingworth
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Michael Fitch
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Nadan Wang
- Cardiology Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mark J Kohr
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States.
| |
Collapse
|
7
|
Carrick-Ranson G, Howden EJ, Brazile TL, Levine BD, Reading SA. Effects of aging and endurance exercise training on cardiorespiratory fitness and cardiac structure and function in healthy midlife and older women. J Appl Physiol (1985) 2023; 135:1215-1235. [PMID: 37855034 PMCID: PMC11918309 DOI: 10.1152/japplphysiol.00798.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 10/05/2023] [Accepted: 10/07/2023] [Indexed: 10/20/2023] Open
Abstract
Cardiovascular disease (CVD) remains the leading cause of morbidity and mortality in women in developed societies. Unfavorable structural and functional adaptations within the heart and central blood vessels with sedentary aging in women can act as the substrate for the development of debilitating CVD conditions such as heart failure with preserved ejection fraction (HFpEF). The large decline in cardiorespiratory fitness, as indicated by maximal or peak oxygen uptake (V̇o2max and V̇o2peak, respectively), that occurs in women as they age significantly affects their health and chronic disease status, as well as the risk of cardiovascular and all-cause mortality. Midlife and older women who have performed structured endurance exercise training for several years or decades of their adult lives exhibit a V̇o2max and cardiac and vascular structure and function that are on par or even superior to much younger sedentary women. Therefore, regular endurance exercise training appears to be an effective preventative strategy for mitigating the adverse physiological cardiovascular adaptations associated with sedentary aging in women. Herein, we narratively describe the aging and short- and long-term endurance exercise training adaptations in V̇o2max, cardiac structure, and left ventricular systolic and diastolic function at rest and exercise in midlife and older women. The role of circulating estrogens on cardiac structure and function is described for consideration in the timing of exercise interventions to maximize beneficial adaptations. Current research gaps and potential areas for future investigation to advance our understanding in this critical knowledge area are highlighted.
Collapse
Affiliation(s)
- Graeme Carrick-Ranson
- Department of Surgery, the University of Auckland, Auckland, New Zealand
- Department of Exercise Sciences, the University of Auckland, Auckland, New Zealand
| | - Erin J Howden
- Human Integrative Physiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Tiffany L Brazile
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, Dallas, Texas, United States
- University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Benjamin D Levine
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, Dallas, Texas, United States
- University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Stacey A Reading
- Department of Exercise Sciences, the University of Auckland, Auckland, New Zealand
| |
Collapse
|
8
|
Taube N, Kabir R, Ebenebe OV, Garbus H, Din SMAE, Illingworth E, Fitch M, Wang N, Kohr MJ. Prenatal Arsenite Exposure Alters Maternal Cardiac Remodeling During Late Pregnancy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.28.559986. [PMID: 37808684 PMCID: PMC10557683 DOI: 10.1101/2023.09.28.559986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Exposure to inorganic arsenic through drinking water is widespread and has been linked to many chronic diseases, including cardiovascular disease. Arsenic exposure has been shown to alter hypertrophic signaling in the adult heart, as well as in-utero offspring development. However, the effect of arsenic on maternal cardiac remodeling during pregnancy has not been studied. As such, there is a need to understand how environmental exposure contributes to adverse pregnancy-related cardiovascular events. This study seeks to understand the impact of trivalent inorganic arsenic exposure during gestation on maternal cardiac remodeling in late pregnancy, as well as offspring outcomes. C57BL/6J mice were exposed to 0 (control), 100 or 1000 µg/L sodium arsenite (NaAsO 2 ) beginning at embryonic day (E) 2.5 and continuing through E17.5. Maternal heart function and size were assessed via transthoracic echocardiography, gravimetric measurement, and histology. Transcript levels of hypertrophic markers were probed via qRT-PCR and confirmed by western blot. Offspring outcomes were assessed through echocardiography and gravimetric measurement. We found that exposure to 1000 µg/L iAs abrogated normal physiologic growth of the maternal heart during late pregnancy and reduced transcript levels of estrogen receptor alpha (ERα), progesterone receptor membrane component 1 (Pgrmc1) and progesterone receptor membrane component 2 (Pgrmc2). Both 100 and 1000 µg/L iAs also reduced transcription of protein kinase B (Akt) and atrial natriuretic peptide (ANP). Akt protein expression was also significantly reduced after 1000 µg/L iAs exposure in the maternal heart with no change in activating phosphorylation. This significant abrogation of maternal cardiac hypertrophy suggests that arsenic exposure during pregnancy can potentially contribute to cardiovascular disease. Taken together, our findings further underscore the importance of reducing arsenic exposure during pregnancy and indicate that more research is needed to assess the impact of arsenic and other environmental exposures on the maternal heart and adverse pregnancy events.
Collapse
|
9
|
Holder ER, Alibhai FJ, Caudle SL, McDermott JC, Tobin SW. The importance of biological sex in cardiac cachexia. Am J Physiol Heart Circ Physiol 2022; 323:H609-H627. [PMID: 35960634 DOI: 10.1152/ajpheart.00187.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cardiac cachexia is a catabolic muscle wasting syndrome observed in approximately 1 in 10 heart failure patients. Increased skeletal muscle atrophy leads to frailty and limits mobility which impacts quality of life, exacerbates clinical care, and is associated with higher rates of mortality. Heart failure is known to exhibit a wide range of prevalence and severity when examined across individuals of different ages and with co-morbidities related to diabetes, renal failure and pulmonary dysfunction. It is also recognized that men and women exhibit striking differences in the pathophysiology of heart failure as well as skeletal muscle homeostasis. Given that both skeletal muscle and heart failure physiology are in-part sex dependent, the diagnosis and treatment of cachexia in heart failure patients may depend on a comprehensive examination of how these organs interact. In this review we explore the potential for sex-specific differences in cardiac cachexia. We summarize advantages and disadvantages of clinical methods used to measure muscle mass and function and provide alternative measurements that should be considered in preclinical studies. Additionally, we summarize sex-dependent effects on muscle wasting in preclinical models of heart failure, disuse, and cancer. Lastly, we discuss the endocrine function of the heart and outline unanswered questions that could directly impact patient care.
Collapse
|
10
|
Rossi BRO, Philbois SV, Maida KD, Sánchez-Delgado JC, Veiga AC, Souza HCD. Role of aerobic physical training on cardiac autonomic and morphophysiological dysfunction in hypertensive rats subjected to ovarian hormone deprivation. Braz J Med Biol Res 2022; 55:e11916. [PMID: 35584451 PMCID: PMC9113528 DOI: 10.1590/1414-431x2022e11916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 03/10/2022] [Indexed: 11/22/2022] Open
Abstract
Here we investigated the effects of physical training on cardiovascular autonomic control and cardiac morphofunctional parameters in spontaneously hypertensive rats (SHRs) subjected to ovarian hormone deprivation. Forty-eight 10-week-old SHRs were divided into two groups: ovariectomized (OVX, n=24) and sham (SHAM, n=24). Half of each group (n=12) was trained by swimming for 12 weeks (OVX-T and SHAM-T). Cardiac morphology and functionality were assessed using echocardiography, and autonomic parameters were assessed using double pharmacological autonomic block, baroreflex sensitivity (BRS), and analyses of heart rate variability (HRV) and blood pressure variability (BPV). Ovariectomy did not influence the cardiac autonomic tonus balance unlike physical training, which favored greater participation of the vagal autonomic tonus. Ovariectomy and aerobic physical training did not modify HRV and BRS, unlike BPV, for which both methods reduced low-frequency oscillations, suggesting a reduction in sympathetic vascular modulation. Untrained ovariectomized animals showed a reduced relative wall thickness (RWT) and increased diastolic and systolic volumes and left ventricular diameters, resulting in increased stroke volume. Trained ovariectomized animals presented reduced posterior wall thickness and RWT as well as increased final diastolic diameter, left ventricular mass, and stroke volume. Ovarian hormone deprivation in SHRs promoted morphofunctional adaptations but did not alter the evaluation of cardiac autonomic parameters. In turn, aerobic physical training contributed to a more favorable cardiac autonomic balance to the vagal autonomic component and promoted morphological adaptations but had little effect on cardiac functionality.
Collapse
Affiliation(s)
- B R O Rossi
- Laboratório de Fisiologia do Exercício e Fisioterapia Cardiovascular, Departamento de Ciência da Saúde, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - S V Philbois
- Laboratório de Fisiologia do Exercício e Fisioterapia Cardiovascular, Departamento de Ciência da Saúde, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - K D Maida
- Laboratório de Fisiologia do Exercício e Fisioterapia Cardiovascular, Departamento de Ciência da Saúde, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - J C Sánchez-Delgado
- Faculty of Physical Culture, Sports and Recreation, University Santo Tomás, Bucaramanga, Colombia
| | - A C Veiga
- Laboratório de Fisiologia do Exercício e Fisioterapia Cardiovascular, Departamento de Ciência da Saúde, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - H C D Souza
- Laboratório de Fisiologia do Exercício e Fisioterapia Cardiovascular, Departamento de Ciência da Saúde, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| |
Collapse
|
11
|
Muehleman DL, Crocini C, Swearingen AR, Ozeroff CD, Leinwand LA. Regression from pathological hypertrophy in mice is sexually dimorphic and stimulus-specific. Am J Physiol Heart Circ Physiol 2022; 322:H785-H797. [PMID: 35302880 PMCID: PMC8993523 DOI: 10.1152/ajpheart.00644.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pathological cardiac hypertrophy is associated with increased morbidity and mortality. Understanding the mechanisms whereby pathological cardiac growth can be reversed could have therapeutic value. Here, we show that pathways leading to regression of pathological cardiac hypertrophy are strongly dependent on the hypertrophic trigger and are significantly modified by sex. Two pathological stimuli causing hypertrophy via distinct pathways were administered to male and female mice: Angiotensin II [Ang II] or Isoproterenol [Iso]. Stimuli were removed after 7 days of treatment, and left ventricles (LV) were studied at 1, 4, and 7 days. Ang II-treated Females did not show regression after stimulus removal. Iso-treated males showed rapid LV hypertrophy regression. Somewhat surprisingly, RNAseq analysis at day 1 after removal of triggers revealed only 45 differentially regulated genes in common among all groups, demonstrating distinct responses. Ingenuity Pathway Analysis predicted strong downregulation of the TGFβ1 pathway in all groups except for Ang II-treated females. Consistently, we found significant downregulation of Smad signaling after stimulus removal including in Ang II-treated females. Additionally, the ERK1/2 pathway was significantly reduced in the groups showing regression. Finally, protein degradation pathways were significantly activated only in Iso-treated males 1 day after stimulus removal. Our data indicate that TGFβ1 downregulation may play a role in the regression of pathological cardiac hypertrophy via downregulation of the ERK1/2 pathway and activation of autophagy and proteasome activity in Iso-treated males. This work highlights that the reversal of pathological hypertrophy does not utilize universal signaling pathways and that sex potently modifies this process.
Collapse
Affiliation(s)
- Deanna L Muehleman
- BioFrontiers Institute University of Colorado Boulder; Department of Molecular and Cellular Development, University of Colorado Boulder, Boulder, Colorado, United States
| | - Claudia Crocini
- BioFrontiers Institute University of Colorado Boulder; Department of Molecular and Cellular Development, University of Colorado Boulder; Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Neuromuscular and Cardiovascular Cell Biology; German Center for Cardiovascular Research (DZHK) Partner Site Berlin, Boulder, Colorado
| | - Alison R Swearingen
- Department of Molecular and Cellular Development, University of Colorado Boulder, Boulder, Colorado, United States
| | - Christopher D Ozeroff
- BioFrontiers Institute University of Colorado Boulder; Department of Molecular and Cellular Development, University of Colorado Boulder, Boulder, Colorado, United States
| | - Leslie A Leinwand
- BioFrontiers Institute University of Colorado Boulder; Department of Molecular and Cellular Development, University of Colorado Boulder, Boulder, Colorado, United States
| |
Collapse
|
12
|
Alhumaid W, Small SD, Kirkham AA, Becher H, Pituskin E, Prado CM, Thompson RB, Haykowsky MJ, Paterson DI. A Contemporary Review of the Effects of Exercise Training on Cardiac Structure and Function and Cardiovascular Risk Profile: Insights From Imaging. Front Cardiovasc Med 2022; 9:753652. [PMID: 35265675 PMCID: PMC8898950 DOI: 10.3389/fcvm.2022.753652] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 01/17/2022] [Indexed: 12/26/2022] Open
Abstract
Exercise is a commonly prescribed therapy for patients with established cardiovascular disease or those at high risk for de novo disease. Exercise-based, multidisciplinary programs have been associated with improved clinical outcomes post myocardial infarction and is now recommended for patients with cancer at elevated risk for cardiovascular complications. Imaging studies have documented numerous beneficial effects of exercise on cardiac structure and function, vascular function and more recently on the cardiovascular risk profile. In this contemporary review, we will discuss the effects of exercise training on imaging-derived cardiovascular outcomes. For cardiac imaging via echocardiography or magnetic resonance, we will review the effects of exercise on left ventricular function and remodeling in patients with established or at risk for cardiac disease (myocardial infarction, heart failure, cancer survivors), and the potential utility of exercise stress to assess cardiac reserve. Exercise training also has salient effects on vascular function and health including the attenuation of age-associated arterial stiffness and thickening as assessed by Doppler ultrasound. Finally, we will review recent data on the relationship between exercise training and regional adipose tissue deposition, an emerging marker of cardiovascular risk. Imaging provides comprehensive and accurate quantification of cardiac, vascular and cardiometabolic health, and may allow refinement of risk stratification in select patient populations. Future studies are needed to evaluate the clinical utility of novel imaging metrics following exercise training.
Collapse
Affiliation(s)
- Waleed Alhumaid
- Division of Cardiology, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | | | - Amy A. Kirkham
- Faculty of Kinesiology, University of Toronto, Toronto, ON, Canada
| | - Harald Becher
- Division of Cardiology, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | - Edith Pituskin
- Faculty of Nursing, College of Health Sciences, University of Alberta, Edmonton, AB, Canada
| | - Carla M. Prado
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Richard B. Thompson
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB, Canada
| | - Mark J. Haykowsky
- Faculty of Nursing, College of Health Sciences, University of Alberta, Edmonton, AB, Canada
| | - D. Ian Paterson
- Division of Cardiology, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
- *Correspondence: D. Ian Paterson
| |
Collapse
|
13
|
Sex-Specific Impacts of Exercise on Cardiovascular Remodeling. J Clin Med 2021; 10:jcm10173833. [PMID: 34501285 PMCID: PMC8432130 DOI: 10.3390/jcm10173833] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/21/2021] [Accepted: 08/21/2021] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular diseases (CVD) remain the leading cause of death in men and women. Biological sex plays a major role in cardiovascular physiology and pathological cardiovascular remodeling. Traditionally, pathological remodeling of cardiovascular system refers to the molecular, cellular, and morphological changes that result from insults, such as myocardial infarction or hypertension. Regular exercise training is known to induce physiological cardiovascular remodeling and beneficial functional adaptation of the cardiovascular apparatus. However, impact of exercise-induced cardiovascular remodeling and functional adaptation varies between males and females. This review aims to compare and contrast sex-specific manifestations of exercise-induced cardiovascular remodeling and functional adaptation. Specifically, we review (1) sex disparities in cardiovascular function, (2) influence of biological sex on exercise-induced cardiovascular remodeling and functional adaptation, and (3) sex-specific impacts of various types, intensities, and durations of exercise training on cardiovascular apparatus. The review highlights both animal and human studies in order to give an all-encompassing view of the exercise-induced sex differences in cardiovascular system and addresses the gaps in knowledge in the field.
Collapse
|
14
|
Alaasam VJ, Liu X, Niu Y, Habibian JS, Pieraut S, Ferguson BS, Zhang Y, Ouyang JQ. Effects of dim artificial light at night on locomotor activity, cardiovascular physiology, and circadian clock genes in a diurnal songbird. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 282:117036. [PMID: 33838441 PMCID: PMC8184626 DOI: 10.1016/j.envpol.2021.117036] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/04/2021] [Accepted: 03/28/2021] [Indexed: 05/17/2023]
Abstract
Artificial light is transforming the nighttime environment and quickly becoming one of the most pervasive pollutants on earth. Across taxa, light entrains endogenous circadian clocks that function to synchronize behavioral and physiological rhythms with natural photoperiod. Artificial light at night (ALAN) disrupts these photoperiodic cues and has consequences for humans and wildlife including sleep disruption, physiological stress and increased risk of cardiovascular disease. However, the mechanisms underlying organismal responses to dim ALAN, resembling light pollution, remain elusive. Light pollution exists in the environment at lower levels (<5 lux) than tested in many laboratory studies that link ALAN to circadian rhythm disruption. Few studies have linked dim ALAN to both the upstream regulators of circadian rhythms and downstream behavioral and physiological consequences. We exposed zebra finches (Taeniopygia gutatta) to dim ALAN (1.5 lux) and measured circadian expression of five pacemaker genes in central and peripheral tissues, plasma melatonin, locomotor activity, and biomarkers of cardiovascular health. ALAN caused an increase in nighttime activity and, for males, cardiac hypertrophy. Moreover, downstream effects were detectable after just short duration exposure (10 days) and at dim levels that mimic the intensity of environmental light pollution. However, ALAN did not affect circulating melatonin nor oscillations of circadian gene expression in the central clock (brain) or liver. These findings suggest that dim ALAN can alter behavior and physiology without strong shifts in the rhythmic expression of molecular circadian pacemakers. Approaches that focus on ecologically-relevant ALAN and link complex biological pathways are necessary to understand the mechanisms underlying vertebrate responses to light pollution.
Collapse
Affiliation(s)
- Valentina J Alaasam
- Department of Biology, University of Nevada, Reno, Reno, NV, USA; Program of Ecology, Evolution, and Conservation Biology, University of Nevada, Reno, Reno, NV, USA.
| | - Xu Liu
- Department of Biology, University of Nevada, Reno, Reno, NV, USA
| | - Ye Niu
- Department of Biology, University of Nevada, Reno, Reno, NV, USA
| | - Justine S Habibian
- Department of Nutrition, University of Nevada, Reno, Reno, NV, USA; Program of Cellular and Molecular Biology, University of Nevada, Reno, Reno, NV, USA
| | - Simon Pieraut
- Department of Biology, University of Nevada, Reno, Reno, NV, USA
| | - Brad S Ferguson
- Department of Nutrition, University of Nevada, Reno, Reno, NV, USA; Center for Biomedical Research Excellence in Molecular and Cellular Signal Transduction in the Cardiovascular System, School of Medicine, University of Nevada, Reno, Reno, NV, USA
| | - Yong Zhang
- Department of Biology, University of Nevada, Reno, Reno, NV, USA
| | - Jenny Q Ouyang
- Department of Biology, University of Nevada, Reno, Reno, NV, USA
| |
Collapse
|
15
|
Adjuvant Hormonotherapy and Cardiovascular Risk in Post-Menopausal Women with Breast Cancer: A Large Population-Based Cohort Study. Cancers (Basel) 2021; 13:cancers13092254. [PMID: 34066685 PMCID: PMC8125834 DOI: 10.3390/cancers13092254] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Whether aromatase inhibitors (AIs) increase the risk of cardiovascular (CV) events, compared to tamoxifen, in women with breast cancer is still debated. We evaluated the association between AI and CV outcomes in a large population-based cohort of breast cancer women. METHODS By using healthcare utilization databases of Lombardy (Italy), we identified women ≥50 years, with new diagnosis of breast cancer between 2009 and 2015, who started adjuvant therapy with either AI or tamoxifen. We estimated the association between exposure to AI and CV outcomes (including myocardial infarction, ischemic stroke, heart failure or any CV event) by a Cox proportional hazard model with inverse probability of treatment and censoring weighting. RESULTS The study cohort included 26,009 women starting treatment with AI and 7937 with tamoxifen. Over a median follow-up of 5.8 years, a positive association was found between AI and heart failure (Hazard Ratio = 1.20, 95% CI: 1.02 to 1.42) and any CV event (1.14, 1.00 to 1.29). The CV risk increased in women with previous CV risk factors, including hypertension, diabetes and dyslipidemia. CONCLUSIONS Adjuvant therapy with AI in breast cancer women aged more than 50 years is associated with increased risk of heart failure and combined CV events.
Collapse
|
16
|
Bjorkman KK, Guess MG, Harrison BC, Polmear MM, Peter AK, Leinwand LA. miR-206 enforces a slow muscle phenotype. J Cell Sci 2020; 133:jcs243162. [PMID: 32620696 PMCID: PMC7438006 DOI: 10.1242/jcs.243162] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 06/25/2020] [Indexed: 12/21/2022] Open
Abstract
Striated muscle is a highly specialized collection of tissues with contractile properties that vary according to functional needs. Although muscle fiber types are established postnatally, lifelong plasticity facilitates stimulus-dependent adaptation. Functional adaptation requires molecular adaptation, which is partially provided by miRNA-mediated post-transcriptional regulation. miR-206 is a muscle-specific miRNA enriched in slow muscles. We investigated whether miR-206 drives the slow muscle phenotype or is merely an outcome. We found that miR-206 expression increases in both physiological (including female sex and endurance exercise) and pathological conditions (muscular dystrophy and adrenergic agonism) that promote a slow phenotype. Consistent with that observation, the slow soleus muscle of male miR-206-knockout mice displays a faster phenotype than wild-type mice. Moreover, left ventricles of male miR-206 knockout mice have a faster myosin profile, accompanied by dilation and systolic dysfunction. Thus, miR-206 appears to be necessary to enforce a slow skeletal and cardiac muscle phenotype and to play a key role in muscle sexual dimorphisms.
Collapse
Affiliation(s)
- Kristen K Bjorkman
- Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., UCB596, Boulder, CO 80303, USA
| | - Martin G Guess
- Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., UCB596, Boulder, CO 80303, USA
| | - Brooke C Harrison
- Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., UCB596, Boulder, CO 80303, USA
| | - Michael M Polmear
- Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., UCB596, Boulder, CO 80303, USA
| | - Angela K Peter
- Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., UCB596, Boulder, CO 80303, USA
| | - Leslie A Leinwand
- Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., UCB596, Boulder, CO 80303, USA
| |
Collapse
|
17
|
Summerhill VI, Moschetta D, Orekhov AN, Poggio P, Myasoedova VA. Sex-Specific Features of Calcific Aortic Valve Disease. Int J Mol Sci 2020; 21:ijms21165620. [PMID: 32781508 PMCID: PMC7460640 DOI: 10.3390/ijms21165620] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/21/2020] [Accepted: 07/31/2020] [Indexed: 01/09/2023] Open
Abstract
Calcific aortic valve disease (CAVD) is the most common valvular heart disease in developed countries predominantly affecting the elderly population therefore posing a large economic burden. It is a gradually progressive condition ranging from mild valve calcification and thickening, without the hemodynamic obstruction, to severe calcification impairing leaflet motion, known as aortic stenosis (AS). The progression of CAVD occurs over many years, and it is extremely variable among individuals. It is also associated with an increased risk of coronary events and mortality. The recent insights into the CAVD pathophysiology included an important role of sex. Accumulating evidence suggests that, in patients with CAVD, sex can determine important differences in the relationship between valvular calcification process, fibrosis, and aortic stenosis hemodynamic severity between men and women. Consequently, it has implications on the development of different valvular phenotypes, left ventricular hypertrophy, and cardiovascular outcomes in men and women. Along these lines, taking into account the sex-related differences in diagnosis, prognosis, and treatment outcomes is of profound importance. In this review, the sex-related differences in patients with CAVD, in terms of pathobiology, clinical phenotypes, and outcomes were discussed.
Collapse
Affiliation(s)
- Volha I. Summerhill
- Department of Basic Research, Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia;
- Correspondence:
| | - Donato Moschetta
- Unit for the Study of Aortic, Valvular and Coronary Pathologies, Monzino Cardiology Center IRCCS, 20138 Milan, Italy; (D.M.); (P.P.); (V.A.M.)
- Department of Pharmacological and Biomolecular Sciences, The University of Milan, 20133 Milan, Italy
| | - Alexander N. Orekhov
- Department of Basic Research, Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia;
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia
| | - Paolo Poggio
- Unit for the Study of Aortic, Valvular and Coronary Pathologies, Monzino Cardiology Center IRCCS, 20138 Milan, Italy; (D.M.); (P.P.); (V.A.M.)
| | - Veronika A. Myasoedova
- Unit for the Study of Aortic, Valvular and Coronary Pathologies, Monzino Cardiology Center IRCCS, 20138 Milan, Italy; (D.M.); (P.P.); (V.A.M.)
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia
| |
Collapse
|
18
|
Lack of sexual dimorphism in a mouse model of isoproterenol-induced cardiac dysfunction. PLoS One 2020; 15:e0232507. [PMID: 32645007 PMCID: PMC7347208 DOI: 10.1371/journal.pone.0232507] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/24/2020] [Indexed: 12/20/2022] Open
Abstract
Sex-related differences in cardiovascular diseases are highly complex in humans and model-dependent in experimental laboratory animals. The objective of this work was to comprehensively investigate key sex differences in the response to acute and prolonged adrenergic stimulation in C57Bl/6NCrl mice. Cardiac function was assessed by trans-thoracic echocardiography before and after acute adrenergic stimulation (a single sub-cutaneous dose of isoproterenol 10 mg/kg) in 15 weeks old male and female C57Bl/6NCrl mice. Thereafter, prolonged adrenergic stimulation was achieved by sub-cutaneous injections of isoproterenol 10 mg/kg/day for 14 days in male and female mice. Cardiac function and morphometry were assessed by trans-thoracic echocardiography on the 15th day. Thereafter, the mice were euthanized, and the hearts were collected. Histopathological analysis of myocardial tissue was performed after staining with hematoxylin & eosin, Masson's trichrome and MAC-2 antibody. Gene expression of remodeling and fibrotic markers was assessed by real-time PCR. Cardiac function and morphometry were also measured before and after isoproterenol 10 mg/kg/day for 14 days in groups of gonadectomized male and female mice and sham-operated controls. In the current work, there were no statistically significant differences in the positive inotropic and chronotropic effects of isoproterenol between male and female C57Bl/6NCrl. After prolonged adrenergic stimulation, there was similar degree of cardiac dysfunction, cardiac hypertrophy, and myocardial fibrosis in male and female mice. Similarly, prolonged isoproterenol administration induced hypertrophic and fibrotic genes in hearts of male and female mice to the same extent. Intriguingly, gonadectomy of male and female mice did not have a significant impact on isoproterenol-induced cardiac dysfunction as compared to sham-operated animals. The current work demonstrated lack of significant sex-related differences in isoproterenol-induced cardiac hypertrophy, dysfunction, and fibrosis in C57Bl/6NCrl mice. This study suggests that female sex may not be sufficient to protect the heart in this model of isoproterenol-induced cardiac dysfunction and underscores the notion that sexual dimorphism in cardiovascular diseases is highly model-dependent.
Collapse
|
19
|
Wu J, Dai F, Li C, Zou Y. Gender Differences in Cardiac Hypertrophy. J Cardiovasc Transl Res 2019; 13:73-84. [PMID: 31418109 DOI: 10.1007/s12265-019-09907-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 08/06/2019] [Indexed: 12/17/2022]
Abstract
Cardiac hypertrophy is an adaptive response to abnormal physiological and pathological stimuli, which can be classified into concentric and eccentric hypertrophy, induced by pressure overload or volume overload, respectively. In both physiological and pathological scenarios, females generally show a more favorable form of hypertrophy compared with their male counterparts. However once established, cardiac hypertrophy is a stronger risk factor for heart failure in females. Pre-menopausal women are better protected against cardiac hypertrophy compared with men, but this protection is abolished following menopause and is partially restored after estrogen replacement therapy. Estrogen exerts its protection by counteracting pro-hypertrophy signaling pathways, whereas androgen mostly plays an opposite role in cardiac hypertrophy. We here summarize the progress in the understanding of sexual dimorphisms in cardiac hypertrophy and highlight recent breakthroughs in the regulatory role of sex hormones and their intricate molecular networks, in order to shed light on gender-oriented therapeutic efficacy for pathological hypertrophy.
Collapse
Affiliation(s)
- Jian Wu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, 180 Feng Lin Road, Shanghai, 200032, China.
| | - Fangjie Dai
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, 180 Feng Lin Road, Shanghai, 200032, China
| | - Chang Li
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, 180 Feng Lin Road, Shanghai, 200032, China
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, 180 Feng Lin Road, Shanghai, 200032, China.
| |
Collapse
|
20
|
Dennis PM, Raghanti MA, Meindl RS, Less E, Henthorn E, Devlin W, Murray S, Meehan T, Kutinsky I, Murphy H. Cardiac disease is linked to adiposity in male gorillas (Gorilla gorilla gorilla). PLoS One 2019; 14:e0218763. [PMID: 31242268 PMCID: PMC6594625 DOI: 10.1371/journal.pone.0218763] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/07/2019] [Indexed: 01/17/2023] Open
Abstract
Cardiac disease is a major cause of morbidity and mortality for adult gorillas. Previous research indicates a sex-based difference with predominantly males demonstrating evidence of left ventricular hypertrophy. To evaluate these findings, we analyzed serum markers with cardiac measures in a large sample of gorillas. The study sample included 44 male and 25 female gorillas housed at American Association of Zoo and Aquariums (AZA)-accredited zoos. Serum samples were collected from fasted gorillas during routine veterinary health exams and analyzed to measure leptin, adiponectin, IGF-1, insulin, ferritin, glucose, triglycerides, and cholesterol. Cardiac ultrasonography via transthoracic echocardiogram was performed simultaneously. Three echocardiographic parameters were chosen to assess cardiac disease according to parameters established for captive lowland gorillas: left ventricular internal diameter, inter-ventricular septum thickness, and left ventricular posterior wall thickness. Our data revealed that high leptin, low adiponectin, and lowered cholesterol were significantly and positively correlated with measures of heart thickness and age in males but not in females. Lowered cholesterol in this population would be categorized as elevated in humans. High leptin and low adiponectin are indicative of increased adiposity and suggests a potential parallel with human obesity and cardiovascular disease in males. Interestingly, while females exhibited increased adiposity with age, they did not progress to cardiac disease.
Collapse
Affiliation(s)
- Patricia M. Dennis
- Cleveland Metroparks Zoo, Cleveland, Ohio, United States of America
- Department of Veterinary Preventive Medicine, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail: (PD); (MAR)
| | - Mary Ann Raghanti
- Department of Anthropology and School of Biomedical Sciences, Kent State University, Kent, Ohio, United States of America
- Brain Health Research Institute, Kent State University, Kent, Ohio, United States of America
- * E-mail: (PD); (MAR)
| | - Richard S. Meindl
- Department of Anthropology and School of Biomedical Sciences, Kent State University, Kent, Ohio, United States of America
| | - Elena Less
- Cleveland Metroparks Zoo, Cleveland, Ohio, United States of America
| | - Eric Henthorn
- Fortis College Cuyahoga Falls, Ohio, United States of America
| | - William Devlin
- Oakland University William Beaumont School of Medicine, Rochester, Michigan, United States of America
- Beaumont Michigan Heart Group, Troy, Michigan, United States of America
| | - Suzan Murray
- Smithsonian Conservation Biology Institute, National Zoological Park, Washington DC, United States of America
| | - Thomas Meehan
- Brookfield Zoo, Brookfield, Illinois, United States of America
| | - Ilana Kutinsky
- Oakland University William Beaumont School of Medicine, Rochester, Michigan, United States of America
- Beaumont Michigan Heart Group, Troy, Michigan, United States of America
| | - Hayley Murphy
- Great Ape Heart Project based at Zoo Atlanta, Zoo Atlanta, Atlanta, Georgia, United States of America
| |
Collapse
|
21
|
Shekarforoush S, Koohpeyma F, Safari F. Alteration at transcriptional level of cardiac renin-angiotensin system by letrozole treatment. Acta Cardiol 2019; 74:109-113. [PMID: 29909753 DOI: 10.1080/00015385.2018.1472840] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 04/24/2018] [Indexed: 01/19/2023]
Abstract
INTRODUCTION The use of aromatase inhibitors (AIs) for breast cancer led to a marked change in ventricular function. Since accumulating evidence indicates that overactivation of the cardiac renin-angiotensin system (RAS) plays an important role in the development of cardiovascular diseases such as hypertrophy and remodelling, we aimed to investigate whether letrozole alters the transcription level of RAS related genes in the cardiac tissue. METHODS Twenty four rats were randomly divided into four groups (n = 6 per group): two groups were letrozole treated (1 and 2 mg/kg/day orally), one group was vehicle treated (DMSO) and one group was the control group without any treatment. 12 weeks after beginning treatment with letrozole, we examined the rate of transcription of renin, angiotensinogen, AngII type 1a and 1b (AT1a and AT1b) and type 2 receptors (AT2) in the rat heart using real-time polymerase chain reaction. RESULTS The cardiac mRNA levels of several components of the RAS in the rats treated with letrozole were significantly increased including AT1a receptor (80%), renin (51%), and angiotensinogen (33%). Though not significant, AT2 receptor levels were observed to decrease with increasing doses of letrozole. CONCLUSIONS Letrozole can induce significant changes in some RAS related genes. These alterations are important to understand the pathways and consequences beyond cardiac events induced by breast cancer treatments.
Collapse
Affiliation(s)
| | - Farhad Koohpeyma
- b Endocrine and Metabolism research center , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Fatemeh Safari
- c Biotechnology Research Center, International Campus , Shahid Sadoughi University of Medical Sciences , Yazd , Iran
- d Department of Physiology, Faculty of Medicine , Shahid Sadoughi University of Medical Sciences , Yazd , Iran
| |
Collapse
|
22
|
Pawlicki P, Hejmej A, Milon A, Lustofin K, Płachno BJ, Tworzydlo W, Gorowska-Wojtowicz E, Pawlicka B, Kotula-Balak M, Bilinska B. Telocytes in the mouse testicular interstitium: implications of G-protein-coupled estrogen receptor (GPER) and estrogen-related receptor (ERR) in the regulation of mouse testicular interstitial cells. PROTOPLASMA 2019; 256:393-408. [PMID: 30187340 PMCID: PMC6510843 DOI: 10.1007/s00709-018-1305-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/24/2018] [Indexed: 05/12/2023]
Abstract
Telocytes (TCs), a novel type of interstitial cells, are involved in tissue homeostasis maintenance. This study aimed to investigate TC presence in the interstitium of mouse testis. Additionally, inactivation of the G-coupled membrane estrogen receptor (GPER) in the testis was performed to obtain insight into TC function, regulation, and interaction with other interstitial cells. Mice were injected with a GPER antagonist (G-15; 50 μg/kg bw), and the GPER-signaling effect on TC distribution, ultrastructure, and function, as well as the interstitial tissue interaction of GPER with estrogen-related receptors (ERRs), was examined. Microscopic observations of TC morphology were performed with the use of scanning and transmission electron microscopes. Telocyte functional markers (CD34; c-kit; platelet-derived growth factor receptors α and β, PDGFRα and β; vascular endothelial growth factor, VEGF; and vimentin) were analyzed by immunohistochemistry/immunofluorescence and Western blot. mRNA expression of CD34 as well as ERR α, β, and γ was measured by qRT-PCR. Relaxin and Ca2+ concentrations were analyzed by immunoenzymatic and colorimetric assays, respectively. For the first time, we reveal the presence of TCs in the interstitium together with the peritubular area of mouse testis. Telocytes were characterized by specific features such as a small cell body and extremely long prolongations, constituting a three-dimensional network mainly around the interstitial cells. Expression of all TC protein markers was confirmed. Based on scanning electron microscopic observation in GPER-blocked testis, groups of TCs were frequently seen. No changes were found in TC ultrastructure in GPER-blocked testis when compared to the control. However, tendency to TC number change (increase) after the blockage was observed. Concomitantly, no changes in mRNA CD34 expression and increase in ERR expression were detected in GPER-blocked testes. In addition, Ca2+ was unchanged; however, an increase in relaxin concentration was observed. Telocytes are an important component of the mouse testicular interstitium, possibly taking part in maintaining its microenvironment as well as contractile and secretory functions (via themselves or via controlling of other interstitial cells). These cells should be considered a unique and useful target cell type for the prevention and treatment of testicular interstitial tissue disorders based on estrogen-signaling disturbances.
Collapse
Affiliation(s)
- Piotr Pawlicki
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| | - Anna Hejmej
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| | - Agnieszka Milon
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| | - Krzysztof Lustofin
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| | - Bartosz J Płachno
- Department of Plant Cytology and Embryology, Institute of Botany, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| | - Waclaw Tworzydlo
- Department of Developmental Biology and Invertebrate Morphology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| | - Ewelina Gorowska-Wojtowicz
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| | - Bernadetta Pawlicka
- Department of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| | - Malgorzata Kotula-Balak
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland.
| | - Barbara Bilinska
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Gronostajowa 9, 30-387, Krakow, Poland
| |
Collapse
|
23
|
Bernasochi GB, Boon WC, Delbridge LMD, Bell JR. The myocardium and sex steroid hormone influences. CURRENT OPINION IN PHYSIOLOGY 2018. [DOI: 10.1016/j.cophys.2018.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
24
|
Bernardo BC, Ooi JYY, Weeks KL, Patterson NL, McMullen JR. Understanding Key Mechanisms of Exercise-Induced Cardiac Protection to Mitigate Disease: Current Knowledge and Emerging Concepts. Physiol Rev 2018; 98:419-475. [PMID: 29351515 DOI: 10.1152/physrev.00043.2016] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The benefits of exercise on the heart are well recognized, and clinical studies have demonstrated that exercise is an intervention that can improve cardiac function in heart failure patients. This has led to significant research into understanding the key mechanisms responsible for exercise-induced cardiac protection. Here, we summarize molecular mechanisms that regulate exercise-induced cardiac myocyte growth and proliferation. We discuss in detail the effects of exercise on other cardiac cells, organelles, and systems that have received less or little attention and require further investigation. This includes cardiac excitation and contraction, mitochondrial adaptations, cellular stress responses to promote survival (heat shock response, ubiquitin-proteasome system, autophagy-lysosomal system, endoplasmic reticulum unfolded protein response, DNA damage response), extracellular matrix, inflammatory response, and organ-to-organ crosstalk. We summarize therapeutic strategies targeting known regulators of exercise-induced protection and the challenges translating findings from bench to bedside. We conclude that technological advancements that allow for in-depth profiling of the genome, transcriptome, proteome and metabolome, combined with animal and human studies, provide new opportunities for comprehensively defining the signaling and regulatory aspects of cell/organelle functions that underpin the protective properties of exercise. This is likely to lead to the identification of novel biomarkers and therapeutic targets for heart disease.
Collapse
Affiliation(s)
- Bianca C Bernardo
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Jenny Y Y Ooi
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Kate L Weeks
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Natalie L Patterson
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| |
Collapse
|
25
|
Saeed A, Kampangkaew J, Nambi V. Prevention of Cardiovascular Disease in Women. Methodist Debakey Cardiovasc J 2018; 13:185-192. [PMID: 29744010 DOI: 10.14797/mdcj-13-4-185] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular diseases are the leading cause of morbidity and mortality among women worldwide. The pathophysiological basis of cardiovascular health among men and women is not identical. This leads to variable cardiovascular responses to stimulus and presentation of cardiovascular disease symptoms, both of which can have a direct effect on treatment outcomes. Traditionally, the enrollment of women in clinical trials has been minimal, resulting in a lack of gender-specific analysis of clinical trial data and, therefore, the absence of concrete risk factor assessment among women. However, scientific progress in the past decade has identified a spectrum of risk factors for cardiovascular diseases that may be specific to women. These risk factors, which may include menopause, hypertensive disease of pregnancy, and depression, confer additional risk in women besides the traditional risk factors. The current state of knowledge and awareness about these risk factors is suboptimal at this time. Therefore, although the treatment of cardiovascular diseases is similar in both genders, appropriate risk stratification may be limited in women compared to men. The purpose of this review is to describe the recent trends in identifying female-specific risk factors for cardiovascular diseases, their utility in risk stratification, and current pharmacological options for women with regard to cardiovascular disease prevention.
Collapse
Affiliation(s)
- Anum Saeed
- BAYLOR COLLEGE OF MEDICINE, HOUSTON, TEXAS
| | | | - Vijay Nambi
- MICHAEL E. DEBAKEY VA MEDICAL CENTER, HOUSTON, TEXAS.,BAYLOR COLLEGE OF MEDICINE, HOUSTON, TEXAS
| |
Collapse
|
26
|
Trexler CL, Odell AT, Jeong MY, Dowell RD, Leinwand LA. Transcriptome and Functional Profile of Cardiac Myocytes Is Influenced by Biological Sex. ACTA ACUST UNITED AC 2018; 10:CIRCGENETICS.117.001770. [PMID: 29030402 PMCID: PMC5679409 DOI: 10.1161/circgenetics.117.001770] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 09/05/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Although cardiovascular disease is the primary killer of women in the United States, women and female animals have traditionally been omitted from research studies. In reports that do include both sexes, significant sexual dimorphisms have been demonstrated in development, presentation, and outcome of cardiovascular disease. However, there is little understanding of the mechanisms underlying these observations. A more thorough understanding of sex-specific cardiovascular differences both at baseline and in disease is required to effectively consider and treat all patients with cardiovascular disease. METHODS AND RESULTS We analyzed contractility in the whole rat heart, adult rat ventricular myocytes (ARVMs), and myofibrils from both sexes of rats and observed functional sex differences at all levels. Hearts and ARVMs from female rats displayed greater fractional shortening than males, and female ARVMs and myofibrils took longer to relax. To define factors underlying these functional differences, we performed an RNA sequencing experiment on ARVMs from male and female rats and identified ≈600 genes were expressed in a sexually dimorphic manner. Further analysis revealed sex-specific enrichment of signaling pathways and key regulators. At the protein level, female ARVMs exhibited higher protein kinase A activity, consistent with pathway enrichment identified through RNA sequencing. In addition, activating the protein kinase A pathway diminished the contractile sexual dimorphisms previously observed. CONCLUSIONS These data support the notion that sex-specific gene expression differences at baseline influence cardiac function, particularly through the protein kinase A pathway, and could potentially be responsible for differences in cardiovascular disease presentation and outcomes.
Collapse
Affiliation(s)
- Christa L Trexler
- From the Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, University of Colorado at Boulder (C.L.T., A.T.O., R.D.D., L.A.L.); and Division of Cardiology, School of Medicine, Anschutz Medical Campus, University of Colorado, Aurora (M.Y.J.)
| | - Aaron T Odell
- From the Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, University of Colorado at Boulder (C.L.T., A.T.O., R.D.D., L.A.L.); and Division of Cardiology, School of Medicine, Anschutz Medical Campus, University of Colorado, Aurora (M.Y.J.)
| | - Mark Y Jeong
- From the Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, University of Colorado at Boulder (C.L.T., A.T.O., R.D.D., L.A.L.); and Division of Cardiology, School of Medicine, Anschutz Medical Campus, University of Colorado, Aurora (M.Y.J.)
| | - Robin D Dowell
- From the Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, University of Colorado at Boulder (C.L.T., A.T.O., R.D.D., L.A.L.); and Division of Cardiology, School of Medicine, Anschutz Medical Campus, University of Colorado, Aurora (M.Y.J.)
| | - Leslie A Leinwand
- From the Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, University of Colorado at Boulder (C.L.T., A.T.O., R.D.D., L.A.L.); and Division of Cardiology, School of Medicine, Anschutz Medical Campus, University of Colorado, Aurora (M.Y.J.).
| |
Collapse
|
27
|
Put "gender glasses" on the effects of phenolic compounds on cardiovascular function and diseases. Eur J Nutr 2018; 57:2677-2691. [PMID: 29696400 DOI: 10.1007/s00394-018-1695-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 04/19/2018] [Indexed: 12/15/2022]
Abstract
INTRODUCTION The influence of sex and gender is particularly relevant in cardiovascular diseases (CVD) as well as in several aspects of drug pharmacodynamics and pharmacokinetics. Anatomical and physiological differences between the sexes may influence the activity of many drugs, including the possibility of their interaction with other drugs, bioactive compounds, foods and beverages. Phenolic compounds could interact with our organism at organ, cellular, and molecular levels triggering a preventive action against chronic diseases, including CVD. RESULTS This article will review the role of sex on the activity of these bioactive molecules, considering the existence of sex differences in oxidative stress. It describes the pharmacokinetics of phenolic compounds, their effects on vessels, on cardiovascular system, and during development, including the role of nuclear receptors and microbiota. CONCLUSIONS Although there is a large gap between the knowledge of the sex differences in the phenolic compounds' activity and safety, and the urgent need for more research, available data underlie the possibility that plant-derived phenolic compounds could differently influence the health of male and female subjects.
Collapse
|
28
|
Barnes JN, Fu Q. Sex-Specific Ventricular and Vascular Adaptations to Exercise. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1065:329-346. [PMID: 30051394 DOI: 10.1007/978-3-319-77932-4_21] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Increasing data suggest that there are sex differences in ventricular and vascular adaptations to aerobic (endurance) exercise, which may be attributed to different physical and physiological features in men and women. Despite that cardiovascular control during acute exercise at the same relative work rate (e.g., the percentage of peak oxygen uptake) appears to be similar between the sexes, women have blunted responses or adaptations to prolonged (e.g., ≥1 year) exercise training compared with men. Currently, there is little evidence to suggest that exercise-induced vascular adaptations are different between men and women. Furthermore, sex differences in skeletal muscle adaptations to exercise, and how this influences cardiovascular function, remain unclear. Identifying potential differences and the mechanisms behind such exercise-induced adaptations is important for the optimization of exercise interventions between men and women across the life span.
Collapse
Affiliation(s)
| | - Qi Fu
- Institute for Exercise and Environmental Medicine, Dallas, TX, USA.,University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
29
|
Luckey SW, Haines CD, Konhilas JP, Luczak ED, Messmer-Kratzsch A, Leinwand LA. Cyclin D2 is a critical mediator of exercise-induced cardiac hypertrophy. Exp Biol Med (Maywood) 2017; 242:1820-1830. [PMID: 28901173 PMCID: PMC5714145 DOI: 10.1177/1535370217731503] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 08/23/2017] [Indexed: 01/19/2023] Open
Abstract
A number of signaling pathways underlying pathological cardiac hypertrophy have been identified. However, few studies have probed the functional significance of these signaling pathways in the context of exercise or physiological pathways. Exercise studies were performed on females from six different genetic mouse models that have been shown to exhibit alterations in pathological cardiac adaptation and hypertrophy. These include mice expressing constitutively active glycogen synthase kinase-3β (GSK-3βS9A), an inhibitor of CaMK II (AC3-I), both GSK-3βS9A and AC3-I (GSK-3βS9A/AC3-I), constitutively active Akt (myrAkt), mice deficient in MAPK/ERK kinase kinase-1 (MEKK1-/-), and mice deficient in cyclin D2 (cyclin D2-/-). Voluntary wheel running performance was similar to NTG littermates for five of the mouse lines. Exercise induced significant cardiac growth in all mouse models except the cyclin D2-/- mice. Cardiac function was not impacted in the cyclin D2-/- mice and studies using a phospho-antibody array identified six proteins with increased phosphorylation (greater than 150%) and nine proteins with decreased phosphorylation (greater than 33% decrease) in the hearts of exercised cyclin D2-/- mice compared to exercised NTG littermate controls. Our results demonstrate that unlike the other hypertrophic signaling molecules tested here, cyclin D2 is an important regulator of both pathologic and physiological hypertrophy. Impact statement This research is relevant as the hypertrophic signaling pathways tested here have only been characterized for their role in pathological hypertrophy, and not in the context of exercise or physiological hypertrophy. By using the same transgenic mouse lines utilized in previous studies, our findings provide a novel and important understanding for the role of these signaling pathways in physiological hypertrophy. We found that alterations in the signaling pathways tested here had no impact on exercise performance. Exercise induced cardiac growth in all of the transgenic mice except for the mice deficient in cyclin D2. In the cyclin D2 null mice, cardiac function was not impacted even though the hypertrophic response was blunted and a number of signaling pathways are differentially regulated by exercise. These data provide the field with an understanding that cyclin D2 is a key mediator of physiological hypertrophy.
Collapse
Affiliation(s)
- Stephen W Luckey
- Department of Molecular, Cellular and Developmental Biology and BioFrontiers Institute University of Colorado at Boulder, Boulder, CO 80309, USA
- Biology Department, Seattle University, Seattle, WA 98122, USA
| | - Chris D Haines
- Department of Molecular, Cellular and Developmental Biology and BioFrontiers Institute University of Colorado at Boulder, Boulder, CO 80309, USA
| | - John P Konhilas
- Department of Molecular, Cellular and Developmental Biology and BioFrontiers Institute University of Colorado at Boulder, Boulder, CO 80309, USA
- Sarver Molecular Cardiovascular Research Program, Department of Physiology, University of Arizona, Tucson, AZ 85724, USA
| | - Elizabeth D Luczak
- Department of Molecular, Cellular and Developmental Biology and BioFrontiers Institute University of Colorado at Boulder, Boulder, CO 80309, USA
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Antke Messmer-Kratzsch
- Department of Molecular, Cellular and Developmental Biology and BioFrontiers Institute University of Colorado at Boulder, Boulder, CO 80309, USA
| | - Leslie A Leinwand
- Department of Molecular, Cellular and Developmental Biology and BioFrontiers Institute University of Colorado at Boulder, Boulder, CO 80309, USA
| |
Collapse
|
30
|
Chronic treatment with angiotensin-converting enzyme inhibitor increases cardiac fibrosis in young rats submitted to early ovarian failure. Auton Neurosci 2017; 206:28-34. [PMID: 28712539 DOI: 10.1016/j.autneu.2017.07.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 07/06/2017] [Accepted: 07/06/2017] [Indexed: 11/21/2022]
Abstract
BACKGROUND We investigated whether the treatment with enalapril maleate, combined with aerobic physical training, promotes positive effects on the autonomic balance, the morphology and the cardiac function in female rats submitted to early ovarian failure. METHODS Thirty-five female Wistar rats, ovariectomized at 10weeks of age, were assigned into Ovariectomized rats (OVX) and Ovariectomized rats treated with enalapril maleate (OVX-EM, 10mg-1·kg-1·d-1) Each group was subdivided into sedentary and trained (aerobic swimming training for 10weeks). All animals were submitted to a) double pharmacological autonomic blockade, b) study of morphology and cardiac function by echocardiography, and c) analysis of cardiac fibrosis. RESULTS The OVX-EM sedentary group showed a significant increase in cardiac fibrosis, relative heart weight, interventricular septum thickness and increased sympathetic participation and reduced participation of the vagal tone in the determination of the basal heart rate when compared to the OVX sedentary group. Physical training reduced cardiac fibrosis in both groups, however, with less intensity in the OVX-EM group. It also increased the absolute and relative heart weight and the end-systolic volume. Finally, the OVX-EM trained group showed higher values for left ventricular end-systolic volume and lower values for ejection fraction and shortening fraction than the sedentary OVX-EM group. CONCLUSION Enalapril maleate exacerbated cardiac fibrosis and increased sympathetic participation in the basal heart rate determination, without significantly affecting the cardiac function. Aerobic physical training did not change the cardiac autonomic control, but reduced cardiac fibrosis and had little effect on the cardiac function.
Collapse
|
31
|
Cooke PS, Nanjappa MK, Ko C, Prins GS, Hess RA. Estrogens in Male Physiology. Physiol Rev 2017; 97:995-1043. [PMID: 28539434 PMCID: PMC6151497 DOI: 10.1152/physrev.00018.2016] [Citation(s) in RCA: 301] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 01/06/2017] [Accepted: 01/17/2017] [Indexed: 02/06/2023] Open
Abstract
Estrogens have historically been associated with female reproduction, but work over the last two decades established that estrogens and their main nuclear receptors (ESR1 and ESR2) and G protein-coupled estrogen receptor (GPER) also regulate male reproductive and nonreproductive organs. 17β-Estradiol (E2) is measureable in blood of men and males of other species, but in rete testis fluids, E2 reaches concentrations normally found only in females and in some species nanomolar concentrations of estrone sulfate are found in semen. Aromatase, which converts androgens to estrogens, is expressed in Leydig cells, seminiferous epithelium, and other male organs. Early studies showed E2 binding in numerous male tissues, and ESR1 and ESR2 each show unique distributions and actions in males. Exogenous estrogen treatment produced male reproductive pathologies in laboratory animals and men, especially during development, and studies with transgenic mice with compromised estrogen signaling demonstrated an E2 role in normal male physiology. Efferent ductules and epididymal functions are dependent on estrogen signaling through ESR1, whose loss impaired ion transport and water reabsorption, resulting in abnormal sperm. Loss of ESR1 or aromatase also produces effects on nonreproductive targets such as brain, adipose, skeletal muscle, bone, cardiovascular, and immune tissues. Expression of GPER is extensive in male tracts, suggesting a possible role for E2 signaling through this receptor in male reproduction. Recent evidence also indicates that membrane ESR1 has critical roles in male reproduction. Thus estrogens are important physiological regulators in males, and future studies may reveal additional roles for estrogen signaling in various target tissues.
Collapse
Affiliation(s)
- Paul S Cooke
- Department of Physiological Sciences, University of Florida, Gainesville, Florida; Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, Illinois; Department of Urology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Manjunatha K Nanjappa
- Department of Physiological Sciences, University of Florida, Gainesville, Florida; Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, Illinois; Department of Urology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - CheMyong Ko
- Department of Physiological Sciences, University of Florida, Gainesville, Florida; Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, Illinois; Department of Urology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Gail S Prins
- Department of Physiological Sciences, University of Florida, Gainesville, Florida; Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, Illinois; Department of Urology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Rex A Hess
- Department of Physiological Sciences, University of Florida, Gainesville, Florida; Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, Illinois; Department of Urology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
32
|
Felix ACS, Dutra SGV, Gastaldi AC, Bonfim PC, Vieira S, de Souza HCD. Physical training promotes similar effects to the blockade of angiotensin-converting enzyme on the cardiac morphology and function in old female rats subjected to premature ovarian failure. Exp Gerontol 2017; 109:90-98. [PMID: 28408160 DOI: 10.1016/j.exger.2017.04.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 04/05/2017] [Accepted: 04/08/2017] [Indexed: 02/07/2023]
Abstract
We investigated the effects of angiotensin-converting enzyme (ACE) inhibition and aerobic physical training on the heart of old female rats (82-wk-old) submitted to premature ovarian failure (10-wk.-old). We used different approaches: morphology and function by echocardiography, reactivity of the coronary bed and left ventricular contractibility (Langendorff Technique). Female Wistar ovariectomized (OVX) rats (n=42) were assigned to one of four groups: OVX, vehicle treated only; OVX-EM, Enalapril Maleate only (EM, 10mg·kg-1·d-1); OVX-T, aerobic trained only; and OVX-EMT, treated with Enalapril Maleate and aerobic trained. Both Enalapril Maleate treatment and aerobic training were done in the last 20weeks of the experimental protocol. When compared to the OVX group, the OVX-EM group showed lower values of wall thickness and left ventricular (LV) mass, lower values of coronary bed reactivity and reduced maximum response of LV contractility to dobutamine, while the OVX-T group showed lower values of LV wall thickness, increase in end-systolic volume, reduced maximum response of LV contractility to dobutamine, and left intraventricular pressure due to increased flow. The combination of treatments (EM and aerobic physical training) did not promote additional important effects on the parameters evaluated. Our results suggest similar beneficial effects of physical training and EM treatment on the morphology and cardiac function in old female rats submitted to premature ovarian failure. Although the causes of these benefits are still unknown, both treatments have promoted a decrease in cardiac contractility, and the reduced β1-adrenergic sensitivity suggests that both treatments may attenuate the sympathetic effect on the heart.
Collapse
Affiliation(s)
- Ana Carolina S Felix
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Sabrina G V Dutra
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Ada C Gastaldi
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Pâmela C Bonfim
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Suenimeire Vieira
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Hugo Celso Dutra de Souza
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
33
|
Regitz-Zagrosek V, Kararigas G. Mechanistic Pathways of Sex Differences in Cardiovascular Disease. Physiol Rev 2017; 97:1-37. [PMID: 27807199 DOI: 10.1152/physrev.00021.2015] [Citation(s) in RCA: 458] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Major differences between men and women exist in epidemiology, manifestation, pathophysiology, treatment, and outcome of cardiovascular diseases (CVD), such as coronary artery disease, pressure overload, hypertension, cardiomyopathy, and heart failure. Corresponding sex differences have been studied in a number of animal models, and mechanistic investigations have been undertaken to analyze the observed sex differences. We summarize the biological mechanisms of sex differences in CVD focusing on three main areas, i.e., genetic mechanisms, epigenetic mechanisms, as well as sex hormones and their receptors. We discuss relevant subtypes of sex hormone receptors, as well as genomic and nongenomic, activational and organizational effects of sex hormones. We describe the interaction of sex hormones with intracellular signaling relevant for cardiovascular cells and the cardiovascular system. Sex, sex hormones, and their receptors may affect a number of cellular processes by their synergistic action on multiple targets. We discuss in detail sex differences in organelle function and in biological processes. We conclude that there is a need for a more detailed understanding of sex differences and their underlying mechanisms, which holds the potential to design new drugs that target sex-specific cardiovascular mechanisms and affect phenotypes. The comparison of both sexes may lead to the identification of protective or maladaptive mechanisms in one sex that could serve as a novel therapeutic target in one sex or in both.
Collapse
Affiliation(s)
- Vera Regitz-Zagrosek
- Institute of Gender in Medicine & Center for Cardiovascular Research, Charite University Hospital, and DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Georgios Kararigas
- Institute of Gender in Medicine & Center for Cardiovascular Research, Charite University Hospital, and DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| |
Collapse
|
34
|
Blenck CL, Harvey PA, Reckelhoff JF, Leinwand LA. The Importance of Biological Sex and Estrogen in Rodent Models of Cardiovascular Health and Disease. Circ Res 2016; 118:1294-312. [PMID: 27081111 DOI: 10.1161/circresaha.116.307509] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 03/21/2016] [Indexed: 01/01/2023]
Abstract
Nearly one-third of deaths in the United States are caused by cardiovascular disease (CVD) each year. In the past, CVD was thought to mainly affect men, leading to the exclusion of women and female animals from clinical studies and preclinical research. In light of sexual dimorphisms in CVD, a need exists to examine baseline cardiac differences in humans and the animals used to model CVD. In humans, sex differences are apparent at every level of cardiovascular physiology from action potential duration and mitochondrial energetics to cardiac myocyte and whole-heart contractile function. Biological sex is an important modifier of the development of CVD with younger women generally being protected, but this cardioprotection is lost later in life, suggesting a role for estrogen. Although endogenous estrogen is most likely a mediator of the observed functional differences in both health and disease, the signaling mechanisms involved are complex and are not yet fully understood. To investigate how sex modulates CVD development, animal models are essential tools and should be useful in the development of therapeutics. This review will focus on describing the cardiovascular sexual dimorphisms that exist both physiologically and in common animal models of CVD.
Collapse
Affiliation(s)
- Christa L Blenck
- From the Department of Molecular, Cellular, and Developmental Biology & BioFrontiers Institute, University of Colorado, Boulder (C.L.B., P.A.H., L.A.L.); and Women's Health Research Center and Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (J.F.R.)
| | - Pamela A Harvey
- From the Department of Molecular, Cellular, and Developmental Biology & BioFrontiers Institute, University of Colorado, Boulder (C.L.B., P.A.H., L.A.L.); and Women's Health Research Center and Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (J.F.R.)
| | - Jane F Reckelhoff
- From the Department of Molecular, Cellular, and Developmental Biology & BioFrontiers Institute, University of Colorado, Boulder (C.L.B., P.A.H., L.A.L.); and Women's Health Research Center and Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (J.F.R.)
| | - Leslie A Leinwand
- From the Department of Molecular, Cellular, and Developmental Biology & BioFrontiers Institute, University of Colorado, Boulder (C.L.B., P.A.H., L.A.L.); and Women's Health Research Center and Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (J.F.R.).
| |
Collapse
|
35
|
Kurokawa J, Sasano T, Kodama M, Li M, Ebana Y, Harada N, Honda SI, Nakaya H, Furukawa T. Aromatase knockout mice reveal an impact of estrogen on drug-induced alternation of murine electrocardiography parameters. J Toxicol Sci 2016; 40:339-48. [PMID: 25972195 DOI: 10.2131/jts.40.339] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Our in vitro characterization showed that physiological concentrations of estrogen partially suppressed the I(Kr) channel current in guinea pig ventricular myocytes and the human ether-a-go-go-related gene (hERG) channel currents in CHO-K1 cells regardless of estrogen receptor signaling and revealed that the partially suppressed hERG currents enhanced the sensitivity to the hERG blocker E-4031. To obtain in vivo proof-of-concept data to support the effects of estrogen on cardiac electrophysiology, we here employed an aromatase knockout mouse as an in vivo estrogen-null model and compared the acute effects of E-4031 on cardiac electrophysiological parameters with those in wild-type mice (C57/BL6J) by recording surface electrocardiogram (ECG). The ablation of circulating estrogens blunted the effects of E-4031 on heart rate and QT interval in mice under a denervation condition. Our result provides in vivo proof of principle and demonstrates that endogenous estrogens increase the sensitivity of E-4031 to cardiac electrophysiology.
Collapse
Affiliation(s)
- Junko Kurokawa
- Department of Bio-Informational Pharmacology, Medical Research Institute, Tokyo Medical Dental University
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kische H, Gross S, Wallaschofski H, Völzke H, Dörr M, Nauck M, Felix SB, Haring R. Serum androgen concentrations and subclinical measures of cardiovascular disease in men and women. Atherosclerosis 2016; 247:193-200. [PMID: 26926599 DOI: 10.1016/j.atherosclerosis.2016.02.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 02/09/2016] [Accepted: 02/17/2016] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Most of the observed associations of androgens and sex hormone-binding globulin (SHBG) with subclinical cardiovascular disease (CVD) stem from selected study samples with immunoassay-based hormone measurements. Thus, we used a large population-based sample with total testosterone (TT) and androstenedione (ASD) concentrations measured by liquid chromatography-tandem mass spectrometry. DESIGN Data of 2140 individuals (mean age: 60,8 years) from the cohort Study of Health in Pomerania were assessed at baseline and 5-year follow-up. METHODS Multivariable regression models were implemented to assess cross-sectional and longitudinal associations of TT, free testosterone (fT), ASD, SHBG and dehydroepiandrosterone-sulphate (DHEAS) with measures of subclinical CVD including intima media thickness (IMT), carotid plaques, left ventricular mass (LVM), fractional shortening (FS), relative wall thickness (RWT), and left ventricular geometry. RESULTS Cross-sectional analyses yielded an association of TT with IMT in women (β-coefficient per log unit increase: 0.02; 95% CI: 0.007; 0.45) and ASD with FS in both sexes (men: β-coefficient: -2.94; 95% CI: -4.75; -1.12; women: β-coefficient: 1.64; 95% CI: 0.55; 2.73). In longitudinal analyses, DHEAS was positively associated with FS change (β-coefficient: 2.34; 95% CI: -0.59; 4.08). In women, SHBG was positively associated with incident plaques (Q1 vs. Q3 (Ref.): β-coefficient: 1.35; 95% CI: 1.04; 1.74). In both sexes, longitudinal analyses showed no consistent association of TT with subclinical CVD. CONCLUSIONS Despite several sex-specific associations of androgens and SHBG with subclinical CVD, the present representative study for the age group ≥45 years among men and women from the general population detected no consistent associations in longitudinal analyses.
Collapse
Affiliation(s)
- Hanna Kische
- Institute of Clinical Chemistry and Laboratory Medicine, Germany.
| | - Stefan Gross
- German Centre for Cardiovascular Research (DZHK), Greifswald, Germany; Department of Cardiology, University Medicine Greifswald, Germany
| | - Henri Wallaschofski
- Institute of Clinical Chemistry and Laboratory Medicine, Germany; German Centre for Cardiovascular Research (DZHK), Greifswald, Germany
| | - Henry Völzke
- German Centre for Cardiovascular Research (DZHK), Greifswald, Germany; Institute for Community Medicine, Germany
| | - Marcus Dörr
- German Centre for Cardiovascular Research (DZHK), Greifswald, Germany; Department of Cardiology, University Medicine Greifswald, Germany
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, Germany; German Centre for Cardiovascular Research (DZHK), Greifswald, Germany
| | - Stephan B Felix
- German Centre for Cardiovascular Research (DZHK), Greifswald, Germany; Department of Cardiology, University Medicine Greifswald, Germany
| | - Robin Haring
- Institute of Clinical Chemistry and Laboratory Medicine, Germany; European University of Applied Sciences, Faculty of Applied Public Health, Rostock, Germany
| |
Collapse
|
37
|
Sivasinprasasn S, Shinlapawittayatorn K, Chattipakorn SC, Chattipakorn N. Estrogenic Impact on Cardiac Ischemic/Reperfusion Injury. J Cardiovasc Transl Res 2016; 9:23-39. [PMID: 26786980 DOI: 10.1007/s12265-016-9675-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 01/07/2016] [Indexed: 11/29/2022]
Abstract
The increase in cardiovascular disease and metabolic syndrome incidence following the onset of menopause has highlighted the role of estrogen as a cardiometabolic protective agent. Specifically regarding the heart, estrogen induced an improvement in cardiac function, preserved calcium homeostasis, and inhibited the mitochondrial apoptotic pathway. The beneficial effects of estrogen in relation to cardiac ischemia/reperfusion (I/R) injury, such as reduced infarction and ameliorated post-ischemic recovery, have also been shown. Nevertheless, controversial findings exist and estrogen therapy is reported to be related to a higher rate of thromboembolic events and atrial fibrillation in post-menopausal women. Therefore, greater clarification is needed to evaluate the exact potential of estrogen use in cases of cardiac I/R injury. This article reviews the effects of estrogen, in both acute and chronic treatment, and collates the studies with regard to their in vivo, in vitro, or clinical trial settings in cases of cardiac I/R injury and myocardial infarction.
Collapse
Affiliation(s)
- Sivaporn Sivasinprasasn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Cardiac Electrophysiology unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,School of Medicine, Mae Fah Luang University, Chiang Rai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Krekwit Shinlapawittayatorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Cardiac Electrophysiology unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.,Department of Oral Biology and Diagnostic Science, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand. .,Cardiac Electrophysiology unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand. .,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
38
|
Felix ACS, Dutra SGV, Tezini GCSV, Simões MV, de Souza HCD. Aerobic physical training increases contractile response and reduces cardiac fibrosis in rats subjected to early ovarian hormone deprivation. J Appl Physiol (1985) 2015; 118:1276-85. [DOI: 10.1152/japplphysiol.00483.2014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 03/17/2015] [Indexed: 11/22/2022] Open
Abstract
We investigated the effects of early ovarian hormone deprivation on the heart and the role of physical training in this condition using different approaches: cardiac autonomic tone, contractility, morphology and function, and cardiac fibrosis. Female Wistar rats ( n = 48) were assigned into two groups: ovariectomized (Ovx; 10-wk-old) and control rats (Sham; 10-wk-old). Each group was further divided into two subgroups, sedentary and trained (aerobic training by swimming for 10 wk). The sedentary groups showed similar cardiac autonomic tone values; however, only the Sham group had an increase in vagal participation for the determination of the basal heart rate after physical training. The contractile responses to cardiac β-agonists of the sedentary groups were similar, including an increased response to a β1-agonist (dobutamine) observed after physical training. The Ovx sedentary group presented changes in cardiac morphology, which resulted in decreases in the ejection fraction, fractional shortening, and cardiac index compared with the Sham sedentary group. Physical training did little to alter these findings. Moreover, histology analysis showed a significant increase in cardiac fibrosis in the sedentary Ovx group, which was not observed in the trained Ovx group. We conclude that early ovarian hormone deprivation in rats impairs autonomic control, cardiac morphology, and cardiac function and increases cardiac fibrosis; however, it does not affect the contractility induced by dobutamine and salbutamol. Furthermore, this model of physical training prevented an increase in fibrosis and promoted an increase in the cardiac contractile response but had little effect on cardiac autonomic control or morphological and functional parameters.
Collapse
Affiliation(s)
- Ana Carolina S. Felix
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; and
| | - Sabrina G. V. Dutra
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; and
| | - Geisa C. S. V. Tezini
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; and
| | - Marcus Vinicius Simões
- Division of Cardiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Hugo Celso Dutra de Souza
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; and
| |
Collapse
|
39
|
Howden EJ, Perhonen M, Peshock RM, Zhang R, Arbab-Zadeh A, Adams-Huet B, Levine BD. Females have a blunted cardiovascular response to one year of intensive supervised endurance training. J Appl Physiol (1985) 2015; 119:37-46. [PMID: 25930024 DOI: 10.1152/japplphysiol.00092.2015] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 04/23/2015] [Indexed: 12/22/2022] Open
Abstract
Cross-sectional studies in athletes suggest that endurance training augments cardiovascular structure and function with apparently different phenotypes in athletic males and females. It is unclear whether the longitudinal response to endurance training leads to similar cardiovascular adaptations between sexes. We sought to determine whether males and females demonstrate similar cardiovascular adaptations to 1 yr of endurance training, matched for training volume and intensity. Twelve previously sedentary males (26 ± 7, n = 7) and females (31 ± 6, n = 5) completed 1 yr of progressive endurance training. All participants underwent a battery of tests every 3 mo to determine maximal oxygen uptake (V̇o2max) and left ventricle (LV) function and morphology (cardiac magnetic resonance imaging). Pulmonary artery catheterization was performed before and after 1 yr of training, and pressure-volume and Starling curves were constructed during decreases (lower-body negative pressure) and increases (saline infusion) in cardiac volume. Males progressively increased V̇o2max, LV mass, and mean wall thickness, before reaching a plateau from month 9 to 12 of training. In contrast, despite exactly the same training, the response in females was markedly blunted, with V̇o2max, LV mass, and mean wall thickness plateauing after only 3 mo of training. The response of LV end-diastolic volume was not influenced by sex (males +20% and females +18%). After training Starling curves were shifted upward and left, but the effect was greatest in males (interaction P = 0.06). We demonstrate for the first time clear sex differences in response to 1 yr of matched endurance training, such that the development of ventricular hypertrophy and increase in V̇o2max in females is markedly blunted compared with males.
Collapse
Affiliation(s)
- Erin J Howden
- Institute for Exercise and Environmental Medicine, Dallas, Texas; and University of Texas Southwestern Medical Center, Dallas, Texas
| | - Merja Perhonen
- Institute for Exercise and Environmental Medicine, Dallas, Texas; and
| | | | - Rong Zhang
- Institute for Exercise and Environmental Medicine, Dallas, Texas; and University of Texas Southwestern Medical Center, Dallas, Texas
| | - Armin Arbab-Zadeh
- Institute for Exercise and Environmental Medicine, Dallas, Texas; and
| | | | - Benjamin D Levine
- Institute for Exercise and Environmental Medicine, Dallas, Texas; and University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
40
|
Tang Z, Wang Y, Zhu X, Ni X, Cong B, Lu J. Reduced expression of CRHR2 and Sp-1 in myocardium of ovariectomized rats is improved by exercise training. Gynecol Endocrinol 2015; 31:742-6. [PMID: 26182188 DOI: 10.3109/09513590.2015.1056729] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Exercise training has been looked on as a non-pharmacologic approach to treating ovariectomy (OVX)-induced dysfunctions. In this study, we investigated whether chronic exercise impacts on expression of urocortins (UCNs) and corticotropin-releasing hormone receptor type 2 (CRHR2) in myocardium of OVX rats. Bilateral OVX or sham-operation was performed under anesthesia. Both groups were then divided into two subgroups, with or without treadmill training for 8 weeks. It was found that OVX as well as exercise did not affect the mRNA levels of UCN, UCN2 and UCN3 in myocardium. OVX caused down-regulation of CRHR2 in myocardium. Exercise training reversed the OVX-induced reduction of CRHR2, but had no influence on CRHR2 level in sham rats. OVX resulted in a decrease in estrogen receptor α (ERα) expression in myocardium, which was restored by exercise. Moreover, exercise training also reversed OVX-induced down-regulation of specific protein-1 (Sp-1) expression in myocardium. CRHR2 expression level correlated with Sp-1 and ERα level in myocardium. These results indicate that exercise training can restore the CRHR2 level in myocardium of OVX rats, which is associated with ERα and Sp-1 expression.
Collapse
Affiliation(s)
- Zhiping Tang
- a School of Kinesiology, Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport , Shanghai , China and
| | - Yujun Wang
- a School of Kinesiology, Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport , Shanghai , China and
| | - Xiaoyan Zhu
- b Department of Physiology , Second Military Medical University , Shanghai , China
| | - Xin Ni
- b Department of Physiology , Second Military Medical University , Shanghai , China
| | - Binhai Cong
- b Department of Physiology , Second Military Medical University , Shanghai , China
| | - Jianqiang Lu
- a School of Kinesiology, Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport , Shanghai , China and
| |
Collapse
|
41
|
Dworatzek E, Mahmoodzadeh S, Schubert C, Westphal C, Leber J, Kusch A, Kararigas G, Fliegner D, Moulin M, Ventura-Clapier R, Gustafsson JA, Davidson MM, Dragun D, Regitz-Zagrosek V. Sex differences in exercise-induced physiological myocardial hypertrophy are modulated by oestrogen receptor beta. Cardiovasc Res 2014; 102:418-28. [DOI: 10.1093/cvr/cvu065] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
|
42
|
Wan R, Jin L, Zhu C, Liu Y, Li L, Guo R, Li S. Sex difference in urocortin production is contributory to the gender disparity in a rat model of vasculitis induced by sodium laurate. Endocrinology 2013; 154:4663-74. [PMID: 24064363 DOI: 10.1210/en.2013-1572] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases, the most common leading death diseases, occur more in men than women of the same ages. Increasing evidence shows that urocortin (Ucn1), an autocrine or paracrine pro-inflammatory factor, can be regulated by sex hormones. The purpose of the study is to investigate the role of Ucn1 in gender disparity in a sodium laurate-induced vasculitis model. Rats exhibited visible signs of vasculitis on the 14th day after sodium laurate injection. Inflammatory states of the rat femoral artery were observed by histological examination. Significant gender disparity, with the symptoms much grosser in males than females, was seen. In males, the serum levels of Ucn1, prostaglandin estradiol, and soluble intercellular adhesion molecule-1 and the expressions of Ucn1, cyclooxygenase-2, and intercellular adhesion molecule-1 in femoral artery were higher than those in females. Orchidectomy significantly ameliorated the symptoms of vasculitis accompanied with a decrease in the plasma Ucn1 level. However, estradiol supplement after orchidectomy failed to improve the inflammatory states further. In females, ovariectomy and/or dihydrotestosterone supplement significantly increased Ucn1 level and exacerbated symptoms of vasculitis. Furthermore, ip administration of rabbit antiserum to Ucn1 almost abolished the gender differences in vasculitis. These results demonstrated that vasculitis of this model is androgen-responsive and hormonal manipulation by surgical orchidectomy could substantially attenuate the symptoms of vasculitis. Moreover, Ucn1 is a contributory factor to the gender disparity in vasculitis and dihydrotestosterone-promoted Ucn1 secretion exacerbated the development of vasculitis.
Collapse
Affiliation(s)
- Rong Wan
- Department of Pharmacology, Nanjing Medical University-Jiangsu Provincial Key Lab of Cardiovascular Diseases and Molecular Intervention, Nanjing, China.
| | | | | | | | | | | | | |
Collapse
|
43
|
Sex differences in exercise-induced cardiac hypertrophy. Pflugers Arch 2013; 465:731-7. [DOI: 10.1007/s00424-013-1225-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 01/25/2013] [Accepted: 01/28/2013] [Indexed: 10/27/2022]
|
44
|
Schulz EM, Correll RN, Sheikh HN, Lofrano-Alves MS, Engel PL, Newman G, Schultz JEJ, Molkentin JD, Wolska BM, Solaro RJ, Wieczorek DF. Tropomyosin dephosphorylation results in compensated cardiac hypertrophy. J Biol Chem 2012; 287:44478-89. [PMID: 23148217 DOI: 10.1074/jbc.m112.402040] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Phosphorylation of tropomyosin (Tm) has been shown to vary in mouse models of cardiac hypertrophy. Little is known about the in vivo role of Tm phosphorylation. This study examines the consequences of Tm dephosphorylation in the murine heart. Transgenic (TG) mice were generated with cardiac specific expression of α-Tm with serine 283, the phosphorylation site of Tm, mutated to alanine. Echocardiographic analysis and cardiomyocyte cross-sectional area measurements show that α-Tm S283A TG mice exhibit a hypertrophic phenotype at basal levels. Interestingly, there are no alterations in cardiac function, myofilament calcium (Ca(2+)) sensitivity, cooperativity, or response to β-adrenergic stimulus. Studies of Ca(2+) handling proteins show significant increases in sarcoplasmic reticulum ATPase (SERCA2a) protein expression and an increase in phospholamban phosphorylation at serine 16, similar to hearts under exercise training. Compared with controls, the decrease in phosphorylation of α-Tm results in greater functional defects in TG animals stressed by transaortic constriction to induce pressure overload-hypertrophy. This is the first study to investigate the in vivo role of Tm dephosphorylation under both normal and cardiac stress conditions, documenting a role for Tm dephosphorylation in the maintenance of a compensated or physiological phenotype. Collectively, these results suggest that modification of the Tm phosphorylation status in the heart, depending upon the cardiac state/condition, may modulate the development of cardiac hypertrophy.
Collapse
Affiliation(s)
- Emily M Schulz
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|